Skip to main content

Nano-immunotherapy: overcoming delivery challenge of immune checkpoint therapy

A Correction to this article was published on 18 November 2023

This article has been updated

Abstract

Immune checkpoint (ICP) molecules expressed on tumor cells can suppress immune responses against tumors. ICP therapy promotes anti-tumor immune responses by targeting inhibitory and stimulatory pathways of immune cells like T cells and dendritic cells (DC). The investigation into the combination therapies through novel immune checkpoint inhibitors (ICIs) has been limited due to immune-related adverse events (irAEs), low response rate, and lack of optimal strategy for combinatorial cancer immunotherapy (IMT). Nanoparticles (NPs) have emerged as powerful tools to promote multidisciplinary cooperation. The feasibility and efficacy of targeted delivery of ICIs using NPs overcome the primary barrier, improve therapeutic efficacy, and provide a rationale for more clinical investigations. Likewise, NPs can conjugate or encapsulate ICIs, including antibodies, RNAs, and small molecule inhibitors. Therefore, combining the drug delivery system (DDS) with ICP therapy could provide a profitable immunotherapeutic strategy for cancer treatment. This article reviews the significant NPs with controlled DDS using current data from clinical and pre-clinical trials on mono- and combination IMT to overcome ICP therapeutic limitations.

Graphical Abstract

Introduction

The immune system is the most powerful arm in the body defense system to fight against tumors [1]. However, the tumor ability to escape this strong response makes cancer a progressive and hard-to-treat disease [2, 3]. Cancer immunotherapy (IMT) that focuses on immunoregulatory factors brings the cancer therapeutic method to another spirit [4,5,6] and includes the antibody(Ab) and cell therapy-based approaches currently under close investigation worldwide [7, 8]. Immune checkpoints (ICPs) are a variety of inhibitory mechanisms that are integrated into the immune system. They are essential for self-tolerance and regulating the latency and intensity of physiological immune responses in peripheral tissues to reduce collateral tissue damage. Tumors can control various immune checkpoint pathways as a primary immune resistance mechanism [9]. Novel ICP receptors, which include programmed death 1 (PD-1) and its ligand (PD-L1), and cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), are shown to suppress T cells presented at the tumor site [10, 11]. ICP inhibitors (ICI) are developed using antibodies (Abs), RNAs, peptides, or small molecules which can block ICP proteins. By the end of 2022, at least seven types of ICIs, including PD-1 inhibitors (Nivolumab, Cemiplimab, Pembrolizumab), PD-L1 inhibitors (Avelumab, Durvalumab, and Atezolizumab) and CTLA-4 inhibitor (Ipilimumab) have been approved by food and drug administration (FDA) for the various cancer therapies [12]. Furthermore, T cell immunoglobulin and mucin-domain containing-3 (TIM-3) [13], Lymphocyte activation gene-3 (LAG-3) [14], T cell surface protein containing an immunoglobulin variable (IgV) domain, a transmembrane domain and an immunoreceptor tyrosine-based inhibitory motif (ITIM), which is called TIGIT (T cell immunoglobulin and ITIM domain) [15], B and T lymphocyte attenuator (BTLA) [16], V-domain immunoglobulin suppressor of T cell activation (VISTA) [17], and B7 homolog 3 protein (B7-H3) [18] are next-generation of ICPs in the tumor microenvironment (TME). The schematic representative interplay of current ICPs with relevant specific ligands and their function on CD8/CD4+ T cells is shown in Fig. 1.

Fig. 1
figure 1

Representative profile of potential ICPs with relevant specific ligands and their function on CD8/CD4+ T cells. The interaction among negative co-inhibitory ICPs on CD8/CD4+ T cells, including TIM-3, PD-1, LAG-3, CTLA-4, VISTA, TIGIT, and BTLA-4 and positive co-inhibitory ICPs interact with TCR, and CD28 with their membrane protein of APC or tumor cells

ICIs can activate systemic immune responses, leading to toxicities and resistance. Recently, ICIs indicate gained considerable attention in cancer therapy due to their exceptional significance in antitumor responses and long-term remissions [19,20,21]. It is no exaggeration to say that ICIs are among the most widely successful immunomodulators developed so far [22, 23]. However, ICIs have some kinds of disadvantages, like inducing numerous-immune related adverse events (irAEs) [24,25,26], disruption of the balance or regulation of immune responses [9], self-tolerance, and normal homeostasis of the immune system [27,28,29]. Thus, ICI therapy can cause myocarditis, autoimmune colitis, vitiligo, psoriasiform dermatitis, hepatitis, neuritis, and endocrinopathies such as type 1 diabetes and pancreatitis [26].

ICIs have limitations that combination therapy is adopted due to their adverse effcts including ICI side effects on human body, irAEs effect on clinical outcomes and ICIs endocrine side effects. In human body issue, ICIs work by releasing the brakes on the immune system, allowing it to attack cancer cells. However, this can also cause the immune system to attack normal cells in the body, leading to irAEs that can affect various organs and tissues.The most important irAEs of ICI therapy is as follows: Skin rash and Pruritus, Vitiligo, Colitis, Hepatitis, Pancreatitis, Myocarditis, Nephritis, Pneumonitis, Hypophysitis, Thyroiditis, Adrenal insufficiency, Type 1 diabetes and Neurological disorders (such as encephalitis, myasthenia gravis, and Guillain–Barre syndrome) [30]. Furethermore in a irAEs effect on clinical outcomes, the relationship between irAEs and clinical outcomes in all solid malignancies treated with ICIs was examined. A systematic review of the literature was conducted, and it was found that the development of irAEs was associated with better objective response rate (ORR), progression-free survival (PFS), and overall survival (OS) in patients with metastatic melanoma, lung cancer, renal cell carcinoma, urothelial cancer, head and neck cancer, and gastrointestinal cancers. It was also noted that grade 3 or 4 irAEs were associated with increased ORR but worse OS. The incidence of irAEs can be considered a predictive biomarker of treatment efficacy and toxicity associated with the use of ICIs, according to the study [31].

Despite the fact that there are many reports of endocrine side effects associated with cancer IMT, it is still not clear what their exact prevalence and mechanism. These adverse events include hypophysitis, thyroid disease, and primary adrenal insufficiency. Hypophysitis is a distinctive side effect of CTLA-4 blocking Abs, and prolonged or lifelong substitutive hormonal treatment is often required. The mechanism of injury to the endocrine system triggered by these drugs is yet to be fully elucidated, and well-designed studies are needed to find and validate predictive factors of autoimmune toxicity [32]. The irAEs associated with ICIs, can be distinct from conventional chemotherapy-related toxicities; this highlights the importance of awareness of the clinical presentation, diagnosis, and management of irAEs. The frequency of irAEs is dependent on the agents used, exposure time, and administered dose but also on the patient's intrinsic risk factors.

In order to reduce the side effects of ICI therapy and increase its therapeutic efficacy, it is essential to develop a drug delivery system (DDS) [33, 34]. To that aim, developing DDS and manipulating nano-biomaterial can help us design smart nano-carriers and overcome this barrier by directing ICIs toward our desirable location, achieving TME remodeling, and boosting anti-tumor immunity, and, subsequently, a safe and efficient cancer IMT [35,36,37,38,39].

Nanoparticles (NPs) emerged as significant tools by providing a targeted approach to effectively delivering cancer drugs. Inherent small size, shape, and flexible preparation of NPs, as well as numerous benefits like improved intracellular infiltration, hydrophobic solubility, reduced nonspecific uptake, and reduced toxicity of cancer therapy, all contribute to their ability to increase the efficacy and overcome the limitations of ICI therapy. Taken together, the superiorities of the combinational approaches with NPs and ICI originated from their ability to carry multiple cargos, protecting from nuclease, controlled release, diminished systemic harmful side effects, and modified pharmacodynamics (PD) effects of the cargos [40, 41]. The key idea is the use of confident vehicles to direct medications toward specific organs and cell types rather than systemic delivery, which is responsible for various off-target consequences. Likewise, the success of various nano-biomaterials as a tremendous carrier of ICI therapy was accurately demonstrated. In addition, it was shown that mono-immunotherapy (mono-IMT) with ICIs leads to higher tumor resistance and limited responses. Mono-IMT is the use of one ICI, which is drug that helps to activate the immune system to attack cancer cells. These drugs target specific proteins on the surface of cancer cells and immune cells, allowing the immune system to better recognize and attack cancer cells. Nano-immunotherapy (nano-IMT) has the potential to significantly improve the effectiveness of immunotherapy (IMT) treatments for cancer and other diseases. Thus, nano-IMT emerged to target different inhibitory factors or simultaneously impact both inhibitory and stimulatory pathways with an efficient delivery system [42,43,44]. Recent studies have shown that standard chemotherapy can improve the immune response to tumors and overcome immunoresistance in the TME. This has led to the idea of combining ICIs with standard chemotherapy as a way to enhance the effectiveness of cancer treatment [45, 46]. This novel approach not only leads to the efficient delivery of ICI with optimized dosage and perfect treatment within the body but also synergizes combinatorial therapy of ICI and other drugs (immune molecules and cells or chemotherapy drugs) due to dominating tumor immune evasion through reducing immunomodulatory agents’ exposure, and development of the unique combination of immunotherapies and improving the targeting efficiency treatment with manipulated and targeted NPs [47, 48]. The current review concentrated on the recent interaction between the NP-based delivery of ICI and its effects on cancer IMT. Indeed, we focused on the investigation that conducted the examination based on NPs and ICI simultaneously. Also, as one of this review aims, the combination of NP-based ICI and other inhibitory or stimulatory factors was discussed.

Inhibitor agents for ICP pathways

Antibody blocking agents

The cell–cell attachment and intracellular signaling cascade would charge T cells’ immunosuppressive features due to FOXP3 overexpression and IL-10, TGF-β production, which are crucial mediators of regulatory molecules in the immune system [2]. The ICP blocking modalities can neutralize the cell–cell attachment and induce apoptosis in ICP-expressing cells. The blocking monoclonal antibodies (mAbs) were the first line of ICP blocking systems. The FDA has approved three significant mAbs, including Nivolumab (Opdivo), Pembrolizumab (Keytruda), and Cemiplimab (Libtayo), against their ICP markers (Table 1) [3, 49]. These drugs can target the PD-1+ T cells and induce apoptosis in these cells. PD-1+ T cell depletion positively correlates with a good prognosis of cancer. The anti-PD-1 (aPD-1) treatment should be followed precisely to prevent the autoimmunity problems induced after continuous administration of the aPD-1 regimen. There are many aPD-1 agents in clinical trials, such as IgG1, ScFV, IgG4, single peptide, and some NP-conjugated formulations. New formulations of aPD-1 improve tumor infiltration of these Abs. The poly lactide-co-glycolide acid (PLGA)-loaded aPD-1 NPs can impact CD40+ and CD11c populations in mice tumor models, impacting dendritic cells (DC) activation, intratumoral interferon-γ (IFN-γ) production, and the tumor burden decrement [6]. Although products related to aPD-1 are expanding, the side effects of continued use of these ICIs have led to a greater focus on the design of treatment regimens based on PD-L1. The essential mAbs targeting PD-L1 known so far are Atezolizumab, Avelumab, and Durvalumab, which have received FDA approval. Finally, in 2020, Nivolumab plus Ipilimumab combined with platinum-based chemotherapy (two cycles) was approved by FDA as first-line treatment for metastatic or recurrent non-small cell lung cancer (NSCLC) [12]. Furthermore, the ongoing marketing products and clinical trials of ICI for aPD-1, antiPDL1(aPD-L1) and antiCTLA-4 (aCTLA-4) mAbs are listed in Table 1.

Table 1 FDA-approved ICI mAbs on the market

Nucleic acid-based blocking agents

ICP silencing via small interfering RNA (siRNA) or other inhibitory miRNAs can alter the expression level of ICPs and reduce the downstream signaling cascade protein function. Some NPs can enhance siRNA delivery into the tumor region and tumor-infiltrating lymphocytes (TILs). Some non-viral vectors, such as lipid-coated calcium phosphate (LCP) NPs, can improve the efficacy of siRNA entrance into TILs [51]. The oligonucleotide carriers should have some properties such as small size, cationic charge, lipophilic or amphipathic tendency, stability, clathrin-based endocytic capacity, and non-immunogenic phenotype. The cationic and polymeric NPs can achieve these properties, but the challenges encountered in manipulating, optimizing, and customized decoration for efficient and targeted delivery.

Furthermore, inhibiting ICPs with siRNA-containing NPs can improve the efficacy of cancer vaccines. In one study, combination therapy based on PD-1 and LAG-3 gene suppression in combination with DC vaccination was found to be a practical approach to breast cancer treatment. However, further studies need to be done [52]. The conjugated NPs for delivery of anti-ICP siRNA, such as PD-1, PD-L1, CTLA-4, and LAG-3 in cancer IMT, were shown in Table 2.

Table 2 The various ICIs and NDDS for cancer IMT using monotherapy and combinatorial therapies

Additionally, plasmid DNA can be used to localize PD-L1 trap protein expression along with siRNAs. Creating PD-L1 traps transiently and locally in the TME is possible by loading the PD-L1 coding plasmid DNA into lipid-protamine-DNA NPs, which can synergize with chemotherapy drugs to inhibit tumor growth [53]. Furthermore, PD-1/PD-L1 gene-editing tools are increasingly being investigated, but there are challenges to delivering these tools safely and effectively during clinical trials. As well they designed an NP delivery system using a low molecular weight PEI lipid coating and a PLGA core that can encapsulate a PD-L1 gRNA-CRISPR/Cas9 plasmid and transfect human U87 glioma cells expressing PD-L1 [54]. Using NPs to introduce a PD-L1 GFP-CRISPR/Cas9 plasmid into human glioma cells might provide a novel IMT platform to treat glioblastoma multiforme [54].

Small molecule blocking agents

Targeted anticancer therapies are dominated by small molecules, while IMT uses antibody-based biologics. The widespread use of mAbs against cell surface markers has led to a greater understanding of immunoregulatory ligand-receptor pairs. [55]. mAbs indicate a significant advantage over small molecules in terms of their technical potential to generate selective drugs against biological targets. In addition to reducing off-target drug events, extensive knowledge of the immunoglobulin framework allows better performance of pharmacokinetic (PK) and pharmacology parameters [56]. However, mAb drugs face their limitations; mAb infusion regimens are less convenient in clinical practice than oral administration of small-molecule-based pills. More importantly, irAEs are more controllable in small molecules than Abs due to their shorter half-life and more effortless dose adjustment [55]. Even though ICI Abs have established themselves as the critical components in IMT, these potential advantages of small molecules over Ab drugs have ignited pharmacological efforts to interfere with the intracellular PD-L1-PD1 axis [55, 57]. Furthermore, increasing understanding of T cell intracellular signaling has revealed several negative feedback loops downstream of TCR engagement that could be targeted to boost antitumor T cell immunity. Mitogen-activated protein kinase1 (MAP4K1), also known as hematopoietic progenitor kinase 1 (HPK1) and diacylglycerol kinases (DGK) are two prominent examples. Negative feedback is also mediated by the tyrosine-protein phosphatase non-receptor type 6 (PTPN6, also known as SHP1) and PTPN22 enzymes, as well as the E3 ubiquitin-protein ligase CBL-B128 [55].

Small molecules have been thought to be incapable of inhibiting the PD-L1-PD1 interaction. Nonetheless, the first oral agents, such as CA-170 and GS-4224, have now entered clinical trials. CA-170 is derived from the amino acid sequence serine-asparagine-threonine, discovered through research using motifs from the PD1 primary sequence. According to the reports, the compound targets PD-L1 and VISTA [58, 59]. Even though small-molecule immuno-oncology drugs are becoming increasingly popular, studies on manipulating NPs to change their pharmacologic properties are lacking.

Combination of ICI therapy with chemotherapy

Single-drug therapy has many disadvantages due to cancer heterogeneity and low efficacy. As a result, a combination therapy containing two or more therapeutic agents has developed. Chemoimmunotherapy (CIT) is a new phrase in immunology and oncology, referring to a new spectrum of combinatory cancer treatments. It is about chemotherapy and IMT [60]. Using mAb as an IMT approach besides chemotherapy can boost patient treatment responses. Therefore, ICI therapy, besides chemotherapies, would be included in some guidelines. Nowadays, drug-containing NPs conjugated with ICIs are one of the most critical approaches to CIT drugs. It is formulated by co-encapsulating the drug and ICI in a liposomal carrier or a cationic polymer for siRNA. It would be designed in the NPs-based DDS (NDDS) [61]. Paclitaxel and Carboplatin plus Ipilimumab have been used for NSCLC patients in phase II clinical trials [62]. Some predominant studies in combination therapy are held on NSCLC, such as KEYNOTE-189 (Pembrolizumab and other chemotherapy regimens like) [63], CheckMate 9-LA (Nivolumab and Ipilimumab) [64], and POSEIDON (Durvalumab) [65]. These studies were a combination of chemotherapy and IMT separately. The future of CIT will lead to NP-loaded drug-containing ICIs. aPD-1 and Cisplatin-NPs prepared in a microneedle can release the Cisplatin labeled with aPD-1. These NPs infiltrate the tumor site, and targeting T cells impacts tumor cells simultaneously [66]. Furthermore, the PD-L1 mAb decorated nano-liposome containing Paclitaxel induces more tumor regression than monotherapy [67]. The pH-sensitive nanomicelles containing paclitaxel and aPD-1 enhance immunogenic cell death while PD-1 blocks solid tumors [68].

In addition to Abs, siRNA can be used in CIT as another type of ICIs. Knocking out key genes involved in apoptotic processes and the cell cycle is one of the essential ways in cancer therapy. Hence, siRNA is a promising candidate for inhibiting tumor development and invasion [69]. Combining siRNA therapy with chemotherapeutic drugs can overcome multidrug resistance and promote apoptosis [70]. The presence of suitable DDS based on NPs is promising to overcome these challenges. Although concomitant delivery of ICI and pharmaceutical payloads with NPs are the most critical challenges of these DDS, these approaches indicate a promising feature of nano-IMT for ICI therapy compared to mono-line therapy approaches.

Recently, an innovative approach to vaccination and IMT using an implantable blood clot scaffold loaded with liposomes-protamine-hyaluronic acid NPs (LPH NPs) containing both a vaccine and siRNA. LPH-siRNA that targets PD-L1 and TIM-3 can reduce immunosuppressive signals in mature DCs and prevent the DCs from expressing a regulatory program in the scaffold. The scaffold is intended to recruit immune cells, particularly DCs, to create a DC-rich environment and enhance the immune response [71].

Incorporation of NPs in ICI therapy

ICI-NPs for tumor therapy

Compared with usual immunotherapeutic methods, ICI therapy notably exhibited numerous advantages. However, the major problem is systemic adverse events that could induce severe or life-threatening problems in some patients. Diarrhea, colitis, and flu-like symptoms are those adverse effects that could be managed, but endocrine-related effects and so on could not. Nanomedicine can help us overcome these shortcomings by providing novel and safe DDS [35]. To that end, the diversity of NPs gives them the ability to carry several therapeutic cargos, such as degradable agents and simultaneous soluble and insoluble drugs [72]. Likewise, we can use higher tolerated drug doses using NDDS due to their controlled release ability and fewer off-target effects [73].

Furthermore, NPs can target the local immune microenvironment instead of systemic impact, which makes the therapeutic method safer [35, 74]. Also, investigations revealed that NPs could penetrate the TME rather than conventional therapies by enhanced permeation and retention effect (EPR), thus accumulating in the TME [75]. Regarding the small size of NPs, they could pass through loose, tight junctions of tumor neo-vessels [76].

In the same way, the potential application of the NDDS can help us diminish toxic irAEs of IMT by the EPR effects and improve the risks of IMT by targeted therapy and systemic exposure reduction [76,77,78]. NDDS allows local delivery of ICIs without off-target exposure, thereby preventing auto-reactive and systemic immune responses. The manner, immunogenicity, and bio-distribution of NPs could be influenced by the size, shape, surface charge, decorating ligand, and density [79]. Overall, studies revealed the NDDSs importance in delivering ICIs into tumor cells, reducing toxic side effects, and improving anti-tumor responses. Several nanomaterials have been used to deliver biopharmaceuticals or IMT agents [73, 80]. In addition to enhancing immune checkpoint therapy, Nanoparticles have also been used for the Management of Immune-Related Adverse Events by ICIs [81]. Approximately two dozen clinically approved therapeutic products have produced nanoparticulate systems such as lipid-based, polymeric, inorganic, and hybrid NPs [80, 82,83,84]. The schematic structure of NPs for delivering ICI containing Ab, nucleic acid, and peptide ICIs for IMT is shown in Fig. 2.

Fig. 2
figure 2

Schematic structure of NPs for delivering ICI Ab, siRNA, plasmid DNA, and peptides for cancer IMT

The lipid vehicles for siRNA are among the newest platforms in gene delivery, which could be decorated as a targeting element, such as Ab labeling. It would prevent off-target involvement in healthy non-related cells or organs. These are bio-conjugated or multifunctional NPs [84]. The cationic NPs such as 1,2-dioleoyl-sn-glycerol-3-phosphate (DOPA) can induce the endosomal escaping and siRNA delivery in the cytoplasm [85], especially DOPA-coated dendrimer-siRNA/pDNA lipoid NPs, and the similar combinations can boost the efficacy of siRNA [86].

Unlike liposomal vehicles, which carry drug cargo in the lipid hydrophobic shell and inner hydrophilic core for taking lipophilic and hydrophilic drugs, respectively [87], polymeric NPs (as micelles and conjugates) comprise a robust and polymer-filled core that is better adapted for use in water-insoluble drugs. Attaching therapeutic agents to water-soluble polymers via a covalent bond is another strategy that enhances the lifetime of systemic circulation of drugs and decreases their exposure to normal tissues [85, 86]. Likewise, poly(ε-caprolactone) (PCL), PLGA, polyethyleneimine (PEI), chitosan, poly amido ethylenimine (PAE), poly(phosphazenes); p(DMAEMA), and poly amino amine (PAA) used as synthetic nano-polymers in the delivery. As the most popular one, PEI is used in gene delivery and increment of oligonucleotide or plasmids stability in the complicated process [88, 89]. PLGA, as an FDA-approved biocompatible and biodegradable polymer, shows non-linear and dose-dependent PK and targeted biodistribution features [61]. The majority performance of PLGA NPs is based on uptaking by DC without any specific character recognition, which is used for antigens, vaccines, and other immunotherapeutic agent delivery [62]. Furthermore, PLGA NPs as the carrier could be more helpful in inhibiting the immune escape of the tumor cells, inducing an antitumor-immune response, and blocking the immune check pathways for T cell activation [63, 65]. Chitosan (CS), as a semi-synthetic polymer, is the most identified polymer-based NPs [66, 90].

Inorganic nanomaterials include mesoporous silica NPs (MSN), and iron oxide NPs, play immune signal delivery due to unique physical features. Despite their immunogenicity, these NPs can enhance immune response, showing immunostimulant and immunosuppressant properties in multiple modes [67, 68, 91]. Hybrid NPs have also recently demonstrated promise for IMT molecules by combining organic compounds (lipids or proteins) with polymers, combining the advantages of tailor-made materials with the following advantages: extended circulation in the bloodstream, high encapsulation with various therapeutic agents, minimal premature leakage, targeted therapy simultaneously, and controlled release kinetics [92, 93].

ICI-NPs for pleural and peritoneal effusion

Recent studies have highlighted the potential of NPs as a means of delivering drugs to the peritoneal cavity, particularly for treating peritoneal carcinomatosis. NPs possess numerous advantageous properties as drug carriers, including increased drug retention, prolonged action duration, and controlled drug release, making them effective vehicles for a variety of drugs, including ICIs [134, 135].

In a recent study, researchers investigated the potential of nano-IMT to treat peritoneal carcinomatosis in a mouse model of ovarian cancer. To deliver anti-PD-L1 to the tumor microenvironment (TME) in the peritoneal cavity, the researchers utilized a NPs-based delivery system known as IPI549@HMP. By selectively targeting tumor-associated macrophages (TAMs) within the peritoneal cavity, which play a critical role in promoting tumor growth and suppressing the immune response, the nanoparticles transported the anti-PD-L1 Abs to the intended site. The study demonstrated that delivering anti-PD-L1 Abs to the TAMs in the peritoneal cavity via the NPs platforms resulted in enhanced anti-tumor activity and improved survival in the mouse model [2]. Although there have been significant advancements in utilizing NPs systems for IMT in the treatment of peritoneal carcinomatosis, it is crucial to gain a more comprehensive understanding of the impact of locoregional therapy on the physiological and immune systems of human hosts [136].

NPs and ICI targeting CTLA-4

As discussed, CTLA-4 is the protein expressed in T cells that can regulate CD28 expression [137]. CTLA-4 binding with CD80 and CD86 on APCs can reduce CD28 expression on T cells and inhibit T cell activation (Fig. 1). Thus, blocking of CTLA-4 may augment T cell function and boost anti-tumoral immune response. Therefore, the ICP, like CTLA-4, could be blocked by regulator Abs or the other inhibitor ligands, which is the origin of the ICI idea [138]. In this regard, several clinical trials were conducted in ICIs. Ipilimumab was the first FDA-approved ICI that could target CTLA-4 in metastatic melanoma [139]. It was demonstrated that CTLA-4 blocking led to an improved immune response by up-regulation of CD4+ T cells and down-regulation of Tregs [140]. Similarly, ICI for PD-1 (Nivolumab and Pembrolizumab) and PD-L1 (Durvalumab and Atezolizumab) are now approved to fight several kinds of tumors [141,142,143].

Rahimian et al. designed an investigation to release aCTLA-4 and anti-CD40 Abs sustainably by utilizing a biodegradable poly (D,L lactic-co-hydroxymethyl glycolic acid (PLHMGA) [126, 144]. The study was designed to evaluate microparticles larger than 10 µm for macrophages taken through the local release. The large-size microparticle escaped from macrophage uptake and concluded that aCTLA-4 receiving microparticles were sustainably released in colon carcinoma of mice and exhibited therapeutic efficacy equal 40% survival rate. Subsequently, that treatment strategy successfully reduced local side effects and serum levels of Abs [126].

Systemic administration of ICIs showed much lower potency against brain tumors than other types.[145,146,147]. However, systemic immune system stimulation caused by the free administration of ICIs might improve glioma-bearing mice's survival [148, 149]. To solve the problem and strengthen IMT, the drug should cross the blood–brain barrier (BBB) and penetrate the tumor. In a study of glioblastoma multiforme, a severe aggressive primary brain tumor [150], poly (β-L-malic acid) (PMLA) is utilized as a carrier for aCTLA-4 and aPD-1 Abs to deliver therapy into tumor cells. It was demonstrated that PMLA-based ICIs could cross the BBB and lead to local immune responses and higher survival of glioblastoma-bearing mice. The local treatment could elevate T cells and macrophage activity and reduce CD4+ FoxP3+ T cells (Tregs) in the tumor. Also, the natural killer (NK) cell population and the production of IL-4, 5, 6, and 10 were increased after therapy. PMLA-based ICIs were distributed in the tumor area but not in other healthy brain sites and inhibiting CTLA-4 and PD-1 in the tumor site. It was shown that PMLA-based aCTLA-4 significantly improved the local immune system in the tumor area and elevated the survival of glioma-bearing mice compared with the free drug [132].

In a study of B16F10 melanoma-bearing mice, researchers used a poly(ethylene glycol)-block-poly(D,L-lactide) (PEG–PLA) and the cationic lipid N,N-bis(2-hydroxyethyl)-N-methyl-N-(2- cholesteryoxycarbonyl-aminoethyl) ammonium bromide (BHEM-Chol) as NPs and CTLA-4-specific siRNA (siCTLA-4) as the ICI. Results showed a significant reduction of CTLA-4 in the activated T cells by administering NP-siCTLA-4. Also, the NP-siCTLA-4 regime, compared with NP-based siRNA-negative control, could induce the stimulation, activation, and proliferation of CD8+ and CD4+T cells remarkably and reduce CD4+ FoxP3+ T cells. In addition, NP-siCTLA-4 had a considerable role in delaying tumor development and increasing mice survival time compared to NP-based siRNA-negative control [124].

It was shown that local delivery of aCTLA-4 Abs through Montanide ISA-51 as a slow-release mechanism in the mice-bearing tumor could provide immune responses (higher CD8+ T cells) tumor eradication in low dosages of the drug. Consequently, this low plasma level of ICI minimized the irAEs as the treatment didn't increase autoAbs levels [125, 151].

In another study, functionalized MSN (FMS) was used as an interactive nano environment, elevated protein activity, and carrier with high protein load [152]. It was demonstrated that aCTLA-4 is loaded in FMS with superhigh density to be released long-lastingly. FMS-aCTLA-4 improved the therapeutic responses in melanoma models considerably compared to the free drug released systematically. Interestingly, the rate and durability of aCTLA-4 could be modified through the changes in functional group types and coverages of FMS [125, 148, 152, 153].

In the colorectal cancer (CRC) study, multifunctional upconversion NPs were utilized. This platform was composed of a photosensitizer chlorin e6 (Ce6), and a toll-like receptor-7 (TLR-7) agonist (imiquimod (R837)) as the immune adjuvant, together with aCTLA-4 was able to target tumor growth effectively. The combination therapy potentially eradicates the primary tumors and plays a vital role in the distant tumors' hindrance. This is while unaccompanied UCNP-Ce6-R837 and/or aCTLA-4 could not eliminate the tumor and had only a partial tumor growth delay. The strategy also provoked memory immunity which could support any possible disease recurrence [82, 154].

Altogether, it was shown that CTLA-4 blockades in each way, e.g., mAb or siRNA with the kind of NPs, can improve the anti-tumor efficacy of therapeutic strategies. In addition, other therapeutic factors showed synergistic effects to boost cancer IMT in combination with CTLA-4 blockades. Besides, the NP-based approach makes CTLA-4 blockades penetrate the tumor site and augment the immune system locally, reducing irAEs.

NPs and ICI targeting PD-1/PD-L1 and IDO

ICI targeting PD-1/PD-L1 using NPs

PD-1, one of the inhibitory molecules expressed on the T cell surface, could bind to PD-L1, a surface molecule of cancer cells, and induce inhibitory function [155]. Typically, the interaction between PD-1 and PD-L1 is necessary to maintain immune homeostasis and tolerance [156, 157]. Despite the beneficial function of the PD-1 and PD-L1 axis in normal conditions, studies have focused on blocking this pathway by using ICIs to inhibit tumor growth and improve cancer IMT [21]. There was an association between PD-1/PD-L1 expression level and poor prognosis of the disease and cancer recurrence, especially in breast cancer [158, 159]. In addition, PD-1 expression increased when the disease turned to the late stage [158]. PD-1 protein expressed on lymphocytes, specifically T cells, binds to PD-L1. The PD-1-PD-L1 expression on the tumor cells leads to the tumor escape from T cell-mediated anti-tumoral responses [138].

In an orthotopic CRC model study, an engineered PD-L1 trap plasmid DNA in lipid-protamine-DNA (LPD) NPs was utilized as the alternative to systemic aPD-L1 mAb therapy. It was demonstrated that the combination therapy of the PD-L1 trap (via fusion) with Oxaliplatin showed an improved anti-tumor efficacy and reduced toxic side effects associated with systemic aPD-L1. This is while aPD-L1 mAb plus Oxaliplatin could induce considerable Th17 accumulation in the spleen compared to the NP-PD-L1 trap [53]. Similarly, in a study of liver metastasis of CRC, lipid-coated calcium-phosphate (LCP) NPs were applied as the carrier of PD-L1 and CXCL12 trap plasmids to deliver into the hepatocyte nucleus. The treatment was shown to remarkably elevate immunotherapeutic factors and reduce immunosuppressive agents' concentration in the liver, thus demonstrating higher efficacy compared with the free drug IMT [119]. LCP NP-based therapy was further studied because of its high biocompatibility, good biodegradability, and low toxicity [51, 160]. They also have excellent activity in endosome escape and release of siRNA [161]. The study was designed to knock down the PD-1 of tumor lymphocytes and PD-L1 of tumor cells or possibly APC through the siRNA system to boost tumor-specific lymphocyte responses in an ex vivo model. According to the results, it was revealed that simultaneous blocking of PD-1 and PD-L1 significantly augmented the breast tumor-specific T cell responses. Thus, it could be helpful in T cell-based therapy for cancer patients. Results also demonstrated the strategy elevation of pro-inflammatory cytokines (PICs) such as IFN-γ and tumor necrosis factor (TNF-α). Also, this NP-based ICI was shown as an effective method for delivering cargo into the TME. They claimed that siRNA-mediated therapy is better than the antibody option (Abs) with a short half-life in multiple treatments [161, 162]. An interesting study compared the efficacy of two different NPs, such as layered double hydroxide (LDH) and LCP, to carry for PD-1 specific siRNA (siPD-1). As data showed, LCP NPs had higher cellular uptake and more potential to silence the PD-1 gene in mouse T cell line EL4 than LDH. Besides, LCP NPs showed significantly reduced PD-1 expression in human ex vivo TIL [163].

A study of gastric cancer that utilized folic acid (FA)-modified PEG-S = S-PEI complexes accompanied by superparamagnetic iron oxide NPs for delivering a PD-L1-specific siRNA (si-PD-L1). FA can bind to folate receptor (FR), which is overexpressed in many cancers, including gastric cancer cells, and can boost the complexity of cellular uptake. The FA-PEG-PEI polymers successfully delivered si-PD-L1, with lower off-target toxicity and considerable cellular uptake. In addition, this regime resulted in PD-L1 downregulation in both mRNA and protein levels, further elevating the cytokine release of cocultured T cells [107].

A study of tumor growth used an NP-based dual-targeting therapeutic strategy for combination IMT. The NPs applied were termed immunoswitch particles because they could switch off the PD-L1 on tumor cells and switch on 4-1BB (a co-stimulatory factor) on CD8+ T cells. The conjugation of Abs and iron-Dex NPs led to a synergy between two IMT approaches and a safer way to be useful at low dosages. In vivo analysis of colon cancer and murine melanoma showed that the NP-based combination dual-targeting therapy had a significant antitumor function. The treatment elevated the number and specificity of anti-tumoral CD8+ T cells and changed the endogenous T cell receptor repertoire. In addition, they demonstrated a more impressive recognition ability to identify tumor antigens. However, the immunoswitch strategy hampered tumor growth significantly compared with soluble aPD-L1 and a4-1BB mAbs. This is while particles themselves no indicate any advantages despite the intratumoral injection. Indeed, the conjugation of Abs and NPs is a necessary option for therapeutic anti-tumor activity. It was also demonstrated that bioactive particles had higher local concentrations and lower off-target exposure [116].

The combination therapy concept was further continued with another model study of the B16-F10 melanoma cells and immune-primed 4T1 breast cancer cells. A dual-immunotherapy NPs (DINP), an aPD-1 antagonist, and an anti-OX40 (aOX40) agonist were used as combination therapy. DINP was make-up by conjugating aPD-1 and aOX40 to maleimide-terminated PEG-PLGA NP through thiol-maleimide chemistry. Data showed that the DINP system combination therapy could induce IMT more effectively than free antibody administration. 83% of cured mice eliminated the tumor recurrence, showing the combination therapy that prolonged anti-tumor immune memory responses. Moreover, DINP combination therapy elevated the frequency of CD8+ T cells, the ratio of CD8+ to Tregs, and the effector to central memory T cells compared with the free antibody-administered group. It was shown that synergistic IMT with DINP promoted T cells' activation compared with free antibody IMT [114].

A study of mice bearing subcutaneous B16F10 melanoma, aPD-1, and Zebularine (Zeb) was investigated as a combination therapy. pH-sensitive CaCO3 NPs received aPD-1 for local controlled release. This NP-based aPD-1 was encapsulated with Zeb into the ROS-responsive hydrogel (Zeb-NP-based aPD-1-Gel). As results showed, this NP-based treatment considerably inhibited the tumor compared with the blank-Gel group via the augmentation of T cell-mediated anti-tumor responses. Furthermore, Zeb-NP-based aPD-1-Gel therapy was concentrated accurately in the target site. Compared with the control group, it was shown to not have off-target toxic side effects in the heart, liver, spleen, lung, and spleen [110].

An investigation of the MC38 model of CRC, aPD-1, in conjugation with PEG-PLGA NPs co-encapsulated with TGFβ inhibitor (SD-208), significantly augmented survival rate compared with free drug. It was shown that this treatment reduced off-target toxic adverse effects as the treatment was released after reaching infiltrated T cells [63].

The aPD-1 and glucose oxidase (GOx) were encapsulated into a microneedle patch containing HA grafted with pH-sensitive Dex NPs to treat the B16F10 mouse melanoma model. This NP-based APD-1 treatment was released sustainably because of the acidic condition of the TME. As a result, the MN-GOx patch, which can deliver aPD-1 (MN-GOx-aPD1), demonstrated continuous tumor suppression. Notably, 40% of mice survived 40 days after the therapy, whereas no one survived in the control group. In addition, the infiltration of CD4+ and CD8+ T cells in the MN-GOx-aPD-1 treated group was considerably higher than the untreated one. Furthermore, the combination of aCTLA-4 and aPD-1, with the help of microneedle as a carrier, had a considerable synergistic improvement compared with free Abs. This combination therapy interestingly led to complete control of melanoma as it showed long-term disease-free survival in 70% of treated mice in 60 days [111].

Another study designed a modified platelet system as a biological carrier with the help of ICI in postsurgical cancer IMT. Based on intrinsic platelet properties and bifunctional maleimide linker assistance, platelets were conjugated with a-PD-L1 (P-aPD-L1) to control postsurgical tumor recurrence and metastasis. As platelets interact with circulating tumor cells, aPD-L1 could target tumor cells in the blood circulation and surgical sites. According to results, P-aPD-L1 treated mice showed a 75% survival rate in 60 days, while no mice of all groups survived more than 30 days. As a result, this treatment strategy remarkably diminished cancer growth and metastasis risk and elevated the postoperative survival rate [164].

An investigation in postoperative tumor relapse utilized inflammation-responsive nano-cocoons as the carrier of aPD-1 as the ICI, along with CpG oligodeoxynucleotides (CpG ODNs) as an immune stimulator [112]. CpG ODNs play their potent immunostimulatory function by triggering TLR9-containing cells, which could boost anti-tumor function [165, 166]. In the B16F10 mouse melanoma models, this NP-based ICI therapy, with the simultaneous help of immunostimulators and immunosuppressive inhibitors, hampered tumor relapse more impressively than free aPD-1 and/or CpG nucleotide treatment. It was also demonstrated that the CpG-based delivery of aPD-1 improved the therapeutic method after fragmentation [112].

In a preclinical allograft pancreatic cancer study [120], a liposome-protamine-DNA (LPD) NP-based therapy was designed to treat the disease with the help of plasmids encoding small trapping proteins targeting PD-L1 and CXCL12 (a major chemokine which could inhibit the T cells infiltration [167]. The therapy regulated the TME and boosted the T cell infiltration. Also, it was shown to diminish the metastasis of the tumor cells remarkably. This local and transient delivery of NP-PD-L1 and CXCL12 was shown to reduce irAEs compared with free drug administration [120, 168].

Another recent study used the NDDS to deliver ICIs into the tumor sites. IO@FuDex, composed of iron oxide NPs, fucoidan, and aldehyde-functionalized Dex, was linked with aPD-L1 as the ICI, and aCD3/aCD28 was used as an immune system activator. This new strategy improved the efficacy of the combination therapy in tumor-bearing mice (breast cancer, lung metastasis, and colon cancer) and reduced the drugs' systemic accumulation. As a result, the median survival of tumor-bearing mice increased from 32 days (free aPD-L1 trial) to 63 days [169].

The therapeutic effects of PDT and ICI as combinational therapy were investigated in another B16-F10 melanoma xenograft tumor model study. A combination of PDT through an acid-activatable versatile micelleplex was incorporated by a siPD-L1. The micelleplex utilized a pH-responsive diblock copolymer poly(ethylene glycol)-block poly(diisopropanol amino ethyl methacrylate-cohydroxyethyl methacrylate) (PEG-b-P(DPA-co-HEA) (PDPA), that grafted with a photosensitizer of Pheophorbide A (PPa), and 1,2-epoxytetradecane alkylated oligoethyleneimine (OEI-C14), which is necessary for siRNA complexation and delivery, and the siPD-L1 to silence the PD-L1 expression on the surface of tumor cell. The frequency of CD8+ TIL and the IFN-γ level was remarkably higher in the combination therapy group than in the control ones. The PDT and PD-L1 blockade therapy showed many anti-tumor responses as this regime eliminated tumors utterly. Also, the method was demonstrated to suppress lung metastasis, which is the event in the B16F10 tumor [102].

Similarly, in another study of the B16F10 melanoma tumor, researchers combined PDT therapy and ICI with a micelleplex-based pH-responsive nanocarrier. The therapeutic method focused on the blockade of PD-1/PD-L1 interaction by a siPD-L1 which enhanced anti-tumor immune response via mitochondria-targeting photosensitizer (MTPP) simultaneously. The micelleplexe was made up of two copolymers, including PEG block conjugated poly(2- (diethylamino) ethyl methacrylate (PDEA) (PEG-CDM-PDEA), linking with a pH-sensitive amide bond from 2-propionic- 3-methylmaleic anhydride (CDM) molecule and PEI conjugated PDEA copolymer (PEI-PDEA) which has a potent affinity to bind siRNA. The mild acidic TME cleaves the amide bonds, leading to PEG layer loss and PEI middle layer exposure. The next steps depend on the low pH of endo-lysosomes, which is the cause of disassembly and cargos release. The released siRNA could silence the PD-L1 gene and reduce immune resistance. This is while the laser irradiation through stacked-up MTPP in the mitochondria induces cell apoptosis. Consequently, the therapeutic strategy improved anti-tumor responses and inhibited melanoma metastasis in mice. Also, the primary treated mice's survival rate was 83% after 30 days, remarkably higher than other groups [103]. PD-L1 mAbs copolymerized with PEG-PCL NPs resulted in enhanced uptake of aPD-L1 by tumor sites of mice models [5]. In addition, The conjugated NP formulations of aPD-L1 based on all-trans retinoic acid (ATRA), PLGA, and PEG have more drug delivery potency and more efficacy [170]. Overall, PD-1/PD-L1 blockade was used to overcome problems associated with monotherapy with ICIs. Based on the results of this study, NP-based PD-1/PD-L1 blockade platforms increased IMT accuracy by a significant margin, reduced toxic off-target effects, and provided an extended immunological memory activity that may prevent tumor recurrence after initial eradication.

ICI targeting ADO-PD/ADO using NPs

Research in the B16F10 melanoma model applied engineered cell membrane-derived nanovesicles which presented PD-1 to improve the cancer IMT. This method showed the ability to disrupt PD-1/PD-L1 interactions by binding to the surface PD-L1 of tumor cells. Furthermore, to increase the therapy’s effectiveness, 1-methyl-DL-tryptophan (1-MT), an indoleamine-2,3-dioxygenase (IDO) inhibitor, was encapsulated into PD-1nanovesicles (NVs) [130]. IDO is a rate-limited enzyme in the catabolism and degradation of tryptophan [171] which could inhibit T cells function [172]. Thus, the therapeutic method could simultaneously hamper the PD-1/PD-L1 pathway and IDO. Consequently, in vivo data showed that the therapeutic strategy boosted tumor-specific immune responses by reducing exhausted CD8+ T cells and improving their anti-tumor effects. The therapy significantly delayed the B16F10 tumor growth in the PD-1 NV-administered group compared with aPD-L1 antibody alone. The PD-1 NVs treatment caused 20% of the mice to survive more than 60 days. This is while NVs alone had no remarkable anti-tumor effects [130].

In a study of 4T1tumor-bearing mice, a new redox-sensitive system named PSSN10 consisted of a POEG hydrophilic block and a PNLG hydrophobic block with some NLG919 motifs was used to co-deliver NLG919 (a nontoxic IDO1-selective inhibitor) as an ICI in combination with DOX as a chemotherapy agent. The PSSN10 prodrug polymers that carry NLG919 showed a self-assembly ability to make nanoscale micelles for DOX loading. As a result, this system led to more combination therapy (NLG919 and DOX) accumulation in the tumor sites. Rapid release of DOX was observed when the carrier got to the tumor site. Beyond the chemotherapy effects of DOX, it could augment tumor antigen presentation and promote anti-tumor immune responses. This is while the covalent linkage of NLG919 and a polymer made the cargo release slower and enhanced immunity for an extended period of time. It could be concluded that DOX/PSSN10 mixed micelles showed a significant tumor-inhibitory effect and higher survival of treated mice than free drugs. This was due to the simultaneous release of NLG919 and DOX at the tumor site [127].

A recent study of B16F10 melanoma investigated the synergistic effect of MN-based transcutaneous delivery of aPD-1 and IDO blockade with 1-MT. This strategy was able to release the therapeutic factors in the TME sustainably locally. As a result, 70% of mice bearing melanoma survived 40 days after the therapy, whereas none survived in the related control group. Consequently, this NP-loaded microneedle considerably improved the therapeutic effects and reduced systemic exposure-related side effects [131].

NPs and emerging ICIs

There has been an increase in interest in emerging ICIs and their combination with other approved IMT drugs in recent years. Recently anti LAG-3 Ab (Relatlimab) has gained FDA approval in combination with nivolumab for melanoma cancer treatment [50]. Here again, NPs can be used for targeted therapy and enhance PK parameters of ICI. One research used trimethyl chitosan-dextran sulfate-lactate (TMC-DS-L) NPs loaded with siRNA molecules to inhibit the PD-1 and LAG-3 expressions.

The inhibition of ICP receptors such as LAG-3 and PD-1 on T cells was investigated in this study in order to increase the efficiency of T cells in response to DC vaccines [52]. T cell immunoglobulin mucin-3 (Tim-3) is a newly discovered immune checkpoint molecule and a promising target for HCC treatment [14, 128]. Song et al. developed a novel pH-triggered drug-eluting NP (CC@SR&SF@PP) for simultaneous administration of Tim-3 siRNA and sorafenib to HCC in vivo. Following pH-triggered sorafenib release from SF@PP NPs, tumor proliferation and angiogenesis were significantly inhibited, resulting in remarkable tumor growth suppression in a mouse hepatoma 22 orthotopic tumor model. As a result, the co-delivery of Tim-3 siRNA and sorafenib via this unique pH-triggered drug-eluting NP improves the anti-tumor effect [128]. We anticipate that these combination treatment methods will have tremendous promise in future clinical uses.

In addition to the conventional use of NPs as a delivery system, they can be used in conjunction with IMT as phototherapy (PT) agents. PT approaches commonly use light stimulation to treat peripheral infections and solid tumors due to synergized therapeutic performance, low toxic side effects, and less pain in patients than chemotherapy and radiotherapy [173, 174]. Furthermore, the studies demonstrated that light stimulation could generate tumor-associated antigens (TAAs) and promote innate immune responses [138]. Likewise, photothermal therapy (PTT) had a function in the induction of immune responses and played an adjuvant role [139]. Despite the tremendous anti-tumor ability of PT there is a need to augment anti-tumor immune responses due to the numerous evasion mechanisms that tumors employ to dampen anti-tumor activities [105]. With this regard, the combination therapy of light-triggered PTT with ICI accompaniment was examined in the primary tumor ablation context. Single-walled carbon nanotubes (SWNTs) were combined with a aCTLA-4 antibody and showed an inhibitory role in developing mouse tumor metastasis. Moreover, the polymer-coated SWNTs not only had a photothermal function for the demolition of the tumor, which further led to the release of TAAs, but also demonstrated immunological adjuvant effects as it could induce the maturation of DCs. With the kinds of aCTLA-4, this combination therapy promoted the T cells infiltration into the tumor site. At the same time, it could override the activities of regulatory T cells in distant tumors. It was demonstrated that in a subcutaneous tumor model and a distant established lung metastasis model, SWNT-based PTT combined with aCTLA-4 hampered the growth of remaining cancer cells [105].

Some PTT besides NPs can synergize ICP inhibitory effects, such as Prussian blue NP (PBNP) conjugated anti-CTLA-4 (aCTLA-4) mAbs [140]. The NP-mediated hyperthermia via aCTLA-4 mAbs can cause to depletion of Tregs in tumor masses [141]. PDT is another PT therapy that applies singlet oxygen (1O2) or other reactive oxygen species (ROS), which are the products of photosensitizer molecules enhancement to a high-energy level by a proper excitation light and kills tumor cells [142, 143]. It was exhibited that small peptides are a promising treatment for the ICIs as they can compete with high ICI therapy costs based on Abs. However, the peptides were shown to have some disadvantages beyond the low cost, like short blood circulation time, serum level instability, and side effects [82, 175]. In a study of murine 4T1 breast cancer and CT26 CRC, PLGA NPs were used to load an APD-1 peptide into a hollow gold nanoshell that induced photothermal ablation (PTA) [122]. PTA is another PT method that utilizes the light-generated heat of a near-infrared laser as a weapon to damage tumor cells [176]. This treatment strategy revealed potent anti-tumor responses, which significantly suppress the growth of colorectal and breast cancer. Furthermore, the slow degradation of PLGA led to a controlled release of a aPD-1 peptide, which made this therapy more effective by concentrating on the TME and blocking PD-1 efficiently [122].

Conclusion

Despite the acceptable number of sufferers from specific cancer types responding to ICI therapy, new combinatorial strategies synergized ICI therapy provide a cure or improve the quality of overall survival, at least due to overcoming primary and adaptive resistance performances. Nano-IMT facilitates therapeutic potency by enhanced sequential monotherapy through targeted site-specific or combination therapy to maximize efficacy and minimize toxicities. Thus, combining ICIs with chemotherapy or immunopharmacuetical agents provides flawless simultaneous delivery for T cell activation, immune cellular and molecular modulation and regulation, and assurance of clinical trials. Our review indicates the incorporation of NPs with ICI therapy postulates a promising way to go by applying lipid, polymeric or hybrid NPs for the antitumor treatment and inorganic and metallic NPs for theranostic and imaging purposes. Despite the increasing development of lipid-based nanoparticles (such as liposomes and LNPs) in clinical trials for delivering RNA and immunotherapeutic agents, their utilization for ICIs has been inadequate. However, lipid-based NPs hold promising potential as carriers for ICIs, particularly for antibody or RNA cargoes aimed at blocking ICP pathways.

In addition, polymer NPs act as pioneer immune-NPs due to the activation of APCs and T cells, promotion of effector cells, ease of preparation, quick manipulation, effective targeting, and FDA approval of polymer NPs expanded use for ICI therapy in laboratory research and clinical trials. Furthermore, recent advances demonstrate that the hybrid NPs (lipid-polymer, polymer–polymer, and inorganic-polymer are the perspectives of combination IMT. Likewise, the next decade of ICI therapy will be established using the development of engineered carriers through targeted NPs delivery and RNA and Ab payloads for solid tumors.

Finally, various combination therapies have been developed using mAbs and other therapeutic agents to enhance the efficacy of ICT. Likewise, combining different mAbs that target different ICPs can lead to a more comprehensive blockade of ICPs, resulting in a stronger anti-tumor immune response. Additionally, combining mAbs with chemotherapy or other targeted therapies can help to overcome tumor resistance and enhance the effectiveness of the treatment. Another approach is the use of siRNA to target specific genes involved in cancer growth and survival which used in combination with monoclonal antibodies or other therapies to further enhance the anti-tumor response. Overall, the development of combination therapies using mAbs and other agents has significantly improved the efficacy of immune checkpoint therapy, and has led to the development of new treatment strategies for a wide range of cancers. Ongoing researches which discussed are focused on identifying the most effective combinations of therapies and optimizing treatment protocols to maximize patient outcomes.

Availability of data and materials

Not Applicable.

Change history

Abbreviations

TCR:

T-cell receptor

MHC-II:

Major histocompatibility class II

PD-1:

Anti-programmed death-1

CTLA-4:

Cytotoxic T-lymphocyte-associated protein 4

LAG-3:

Lymphocyte‐associated gene 3

CD:

Cluster of differentiation

VISTA9:

V-domain Ig suppressor of T cell activation

BTLA-4:

B and T lymphocyte attenuator 4

GAL-9:

Galectin-9

HMGB-1:

High mobility group box 1

PtdSer:

Phosphatidylserine

CEACAM-1:

Carcinoembryonic antigen-related cell adhesion molecule 1

LSECtin:

Liver and lymph node sinusoidal endothelial cell C-type lectin

PSGL-1:

P-selectin glycoprotein ligand-1

HVEM:

Herpes virus entry mediator

Necl-5:

Nectin-like molecule-5

FGL-1:

Fibrinogen-like protein 1

References

  1. Swann JB, Smyth MJ. Immune surveillance of tumors. J Clin Investig. 2007;117(5):1137–46.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Mandal CC, Mehta J, Prajapati VK. Programmed Death 1 (PD 1)-mediated T-Cell apoptosis and cancer immunotherapy. Apoptosis Beyond Many Ways Cells Die. 2018. https://doi.org/10.1002/9781119432463.ch29.

    Article  Google Scholar 

  3. Hargadon KM, Johnson CE, Williams CJ. Immune checkpoint blockade therapy for cancer: an overview of FDA-approved immune checkpoint inhibitors. Int Immunopharmacol. 2018;62:29–39.

    CAS  PubMed  Google Scholar 

  4. Ordikhani F, et al. Targeting antigen-presenting cells by anti–PD-1 nanoparticles augments antitumor immunity. JCI insight. 2018. https://doi.org/10.1172/jci.insight.122700.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Xu S, et al. PD-L1 monoclonal antibody-conjugated nanoparticles enhance drug delivery level and chemotherapy efficacy in gastric cancer cells. Int J Nanomed. 2019;14:17.

    CAS  Google Scholar 

  6. Guo L, Zhang H, Chen B. Nivolumab as programmed Death-1 (PD-1) inhibitor for targeted immunotherapy in tumor. J Cancer. 2017;8(3):410–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. June CH, et al. CAR T cell immunotherapy for human cancer. Science. 2018;359(6382):1361–5.

    CAS  PubMed  Google Scholar 

  8. Kimiz-Gebologlu I, Gulce-Iz S, Biray-Avci C. Monoclonal antibodies in cancer immunotherapy. Mol Biol Rep. 2018;45(6):2935–40.

    CAS  PubMed  Google Scholar 

  9. Lee L, Gupta M, Sahasranaman S. Immune Checkpoint inhibitors: An introduction to the next-generation cancer immunotherapy. J Clin Pharmacol. 2016;56(2):157–69.

    CAS  PubMed  Google Scholar 

  10. Qin S, et al. Novel immune checkpoint targets: moving beyond PD-1 and CTLA-4. Mol Cancer. 2019;18(1):1–14.

    CAS  Google Scholar 

  11. Buchbinder EI, Desai A. CTLA-4 and PD-1 pathways: similarities, differences, and implications of their inhibition. Am J Clin Oncol. 2016;39(1):98.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Vaddepally RK, et al. Review of indications of FDA-approved immune checkpoint inhibitors per NCCN guidelines with the level of evidence. Cancers. 2020. https://doi.org/10.3390/cancers12030738.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Andrews LP, et al. LAG 3 (CD 223) as a cancer immunotherapy target. Immunol Rev. 2017;276(1):80–96.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Monney L, et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature. 2002;415(6871):536–41.

    CAS  PubMed  Google Scholar 

  15. Yu X, et al. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat Immunol. 2009;10(1):48.

    CAS  PubMed  Google Scholar 

  16. Yu X, et al. BTLA/HVEM signaling: milestones in research and role in chronic hepatitis B virus infection. Front Immunol. 2019;10:617.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Wang L, et al. VISTA, a novel mouse Ig superfamily ligand that negatively regulates T cell responses. J Exp Med. 2011;208(3):577–92.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Chapoval AI, et al. B7–H3: a costimulatory molecule for T cell activation and IFN-γ production. Nat Immunol. 2001;2(3):269–74.

    CAS  PubMed  Google Scholar 

  19. Siefker-Radtke A, Curti B. Immunotherapy in metastatic urothelial carcinoma: focus on immune checkpoint inhibition. Nat Rev Urol. 2018;15(2):112.

    CAS  PubMed  Google Scholar 

  20. Minn AJ, Wherry EJ. Combination cancer therapies with immune checkpoint blockade: convergence on interferon signaling. Cell. 2016;165(2):272–5.

    CAS  PubMed  Google Scholar 

  21. Fan Q, et al. Toward biomaterials for enhancing immune checkpoint blockade therapy. Adv Func Mater. 2018;28(37):1802540.

    Google Scholar 

  22. Robert C. A decade of immune-checkpoint inhibitors in cancer therapy. Nat Commun. 2020;11(1):1–3.

    Google Scholar 

  23. Bagchi S, Yuan R, Engleman EG. Immune checkpoint inhibitors for the treatment of cancer: clinical impact and mechanisms of response and resistance. Annu Rev Pathol. 2021;16(1):223–49.

    CAS  PubMed  Google Scholar 

  24. Baxi S, et al. Immune-related adverse events for anti-PD-1 and anti-PD-L1 drugs: systematic review and meta-analysis. BMJ. 2018. https://doi.org/10.1136/bmj.k793.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Khan S, Gerber DE. Autoimmunity, checkpoint inhibitor therapy and immune-related adverse events: a review. Seminars Cancer Biol. 2020. https://doi.org/10.1016/j.semcancer.2019.06.012.

    Article  Google Scholar 

  26. Okiyama N, Tanaka R. Immune-related adverse events in various organs caused by immune checkpoint inhibitors. Allergol Int. 2022;71(2):169–78.

    CAS  PubMed  Google Scholar 

  27. Michot J, et al. Immune-related adverse events with immune checkpoint blockade: a comprehensive review. Eur J Cancer. 2016;54:139–48.

    CAS  PubMed  Google Scholar 

  28. Thangavelu G, et al. Programmed death-1 is required for systemic self-tolerance in newly generated T cells during the establishment of immune homeostasis. J Autoimmun. 2011;36(3–4):301–12.

    CAS  PubMed  Google Scholar 

  29. Rivera IO, et al. PD-L1/PD-1 immunotherapy modulates effector T cells homeostasis and function in murine pancreatic cancer. J Immunol. 2016. https://doi.org/10.4049/jimmunol.196.Supp.72.11.

    Article  PubMed  Google Scholar 

  30. Martins F, et al. Adverse effects of immune-checkpoint inhibitors: epidemiology, management and surveillance. Nat Rev Clin Oncol. 2019;16(9):563–80.

    CAS  PubMed  Google Scholar 

  31. Hussaini S, et al. Association between immune-related side effects and efficacy and benefit of immune checkpoint inhibitors—a systematic review and meta-analysis. Cancer Treat Rev. 2021;92: 102134.

    CAS  PubMed  Google Scholar 

  32. Corsello SM, et al. Endocrine side effects induced by immune checkpoint inhibitors. J Clin Endocrinol Metab. 2013;98(4):1361–75.

    CAS  PubMed  Google Scholar 

  33. Lamichhane P, et al. Novel delivery systems for checkpoint inhibitors. Medicines. 2019;6(3):74.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Francis DM, Thomas SN. Progress and opportunities for enhancing the delivery and efficacy of checkpoint inhibitors for cancer immunotherapy. Adv Drug Deliv Rev. 2017;114:33–42.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Musetti S, Huang L. Nanoparticle-mediated remodeling of the tumor microenvironment to enhance immunotherapy. ACS Nano. 2018;12(12):11740–55.

    CAS  PubMed  Google Scholar 

  36. Song W, Musetti SN, Huang L. Nanomaterials for cancer immunotherapy. Biomaterials. 2017;148:16–30.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Hu X, et al. Nanotechnology based therapeutic modality to boost anti-tumor immunity and collapse tumor defense. J Control Release. 2017;256:26–45.

    CAS  PubMed  Google Scholar 

  38. Kelly SH, et al. Biomaterial strategies for generating therapeutic immune responses. Adv Drug Deliv Rev. 2017;114:3–18.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Xie YQ, Wei L, Tang L. Immunoengineering with biomaterials for enhanced cancer immunotherapy. Wiley Interdisciplinary Rev Nanomed Nanobiotechnol. 2018;10(4): e1506.

    Google Scholar 

  40. Ma Q, et al. Synthetic poly (L-glutamic acid)-conjugated CpG exhibits antitumor efficacy with increased retention in tumor and draining lymph nodes after intratumoral injection in a mouse model of melanoma. J Immunother. 2017;40(1):11.

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Ott PA, et al. Combination immunotherapy: a road map. J Immunother Cancer. 2017;5(1):1–15.

    Google Scholar 

  42. Lahori DG, Varamini P. Nanotechnology-based platforms to improve immune checkpoint blockade efficacy in cancer therapy. Future Oncol. 2021;17(6):711–22.

    Google Scholar 

  43. Yu Z, et al. Combined application of nanotechnology and multiple therapies with tumor immune checkpoints. ChemistrySelect. 2020;5(47):14943–54.

    CAS  Google Scholar 

  44. Koshy ST, Mooney DJ. Biomaterials for enhancing anti-cancer immunity. Curr Opin Biotechnol. 2016;40:1–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Principe N, et al. Comprehensive testing of chemotherapy and immune checkpoint blockade in preclinical cancer models identifies additive combinations. Front Immunol. 2022;13: 872295.

    PubMed  PubMed Central  Google Scholar 

  46. Walsh RJ, Sundar R, Lim JSJ. Immune checkpoint inhibitor combinations-current and emerging strategies. Br J Cancer. 2023;128(8):1415–7.

    PubMed  Google Scholar 

  47. Liu Q, et al. Nano-immunotherapy: unique mechanisms of nanomaterials in synergizing cancer immunotherapy. Nano Today. 2021;36: 101023.

    CAS  Google Scholar 

  48. Guevara ML, Persano F, Persano S. Nano-immunotherapy: overcoming tumour immune evasion. Seminars Cancer Biol. 2019. https://doi.org/10.1016/j.semcancer.2019.11.010.

    Article  Google Scholar 

  49. Larkins E, et al. FDA approval summary: pembrolizumab for the treatment of recurrent or metastatic head and neck squamous cell carcinoma with disease progression on or after platinum-containing chemotherapy. Oncologist. 2017;22(7):873.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Paik J. Nivolumab plus relatlimab: first approval. Drugs. 2022;82(8):925–31.

    CAS  PubMed  Google Scholar 

  51. Wu Y, et al. Silencing PD-1 and PD-L1 with nanoparticle-delivered small interfering RNA increases cytotoxicity of tumor-infiltrating lymphocytes. Nanomedicine. 2019;14(8):955–67.

    CAS  PubMed  Google Scholar 

  52. Barshidi A, et al. Dual Blockade of PD-1 and LAG3 immune checkpoints increases dendritic cell vaccine mediated T cell responses in breast cancer model. Pharm Res. 2022;39(8):1851–66.

    CAS  PubMed  Google Scholar 

  53. Song W, et al. Synergistic and low adverse effect cancer immunotherapy by immunogenic chemotherapy and locally expressed PD-L1 trap. Nat Commun. 2018;9(1):1–11.

    Google Scholar 

  54. Tran A, Fierro J, Dou H. CSIG-11 targeting PD-L1 IN glioblastoma using nanoparticle-based gene editing. Neuro-Oncol. 2020. https://doi.org/10.1093/neuonc/noaa215.123.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Offringa R, et al. The expanding role for small molecules in immuno-oncology. Nat Rev Drug Discovery. 2022;21(11):821–40.

    CAS  PubMed  Google Scholar 

  56. Wu Q, et al. Small-molecule inhibitors, immune checkpoint inhibitors, and more: FDA-approved novel therapeutic drugs for solid tumors from 1991 to 2021. J Hematol Oncol. 2022. https://doi.org/10.1186/s13045-022-01362-9.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Sasikumar PG, Ramachandra M. Small-Molecule immune checkpoint inhibitors targeting PD-1/PD-L1 and other emerging checkpoint pathways. BioDrugs. 2018;32(5):481–97.

    CAS  PubMed  Google Scholar 

  58. Sasikumar PG, et al. PD-1 derived CA-170 is an oral immune checkpoint inhibitor that exhibits preclinical anti-tumor efficacy. Commun Biol. 2021;4(1):1–12.

    Google Scholar 

  59. Radhakrishnan V, et al. Excellent CBR and prolonged PFS in non-squamous NSCLC with oral CA-170, an inhibitor of VISTA and PD-L1. Ann Oncol. 2019;30: v494.

    Google Scholar 

  60. Ragelle H, et al. Chitosan nanoparticles for SiRNA delivery in vitro. Methods Mol Biol. 2016;1364:143–50.

    CAS  PubMed  Google Scholar 

  61. Makadia HK, Siegel SJ. Poly lactic-co-glycolic acid (PLGA) as biodegradable controlled drug delivery carrier. Polymers. 2011;3(3):1377–97.

    CAS  PubMed  Google Scholar 

  62. Surendran SP, et al. Bioactive nanoparticles for cancer immunotherapy. Int J Mol Sci. 2018. https://doi.org/10.3390/ijms19123877.

    Article  PubMed  Google Scholar 

  63. Schmid D, et al. T cell-targeting nanoparticles focus delivery of immunotherapy to improve antitumor immunity. Nat Commun. 2017;8(1):1747.

    PubMed  PubMed Central  Google Scholar 

  64. Paz-Ares L, et al. First-line nivolumab plus ipilimumab combined with two cycles of chemotherapy in patients with non-small-cell lung cancer (CheckMate 9LA): an international, randomised, open-label, phase 3 trial. Lancet Oncol. 2021;22(2):198–211.

    CAS  PubMed  Google Scholar 

  65. Luo L, et al. Laser immunotherapy in combination with perdurable PD-1 blocking for the treatment of metastatic tumors. ACS Nano. 2018;12(8):7647–62.

    CAS  PubMed  Google Scholar 

  66. Ghasemi-Chaleshtari M, et al. Concomitant blockade of A2AR and CTLA-4 by siRNA-loaded polyethylene glycol-chitosan-alginate nanoparticles synergistically enhances antitumor T-cell responses. J Cell Physiol. 2020;235(12):10068–80.

    CAS  PubMed  Google Scholar 

  67. Hess KL, Medintz IL, Jewell CM. Designing inorganic nanomaterials for vaccines and immunotherapies. Nano Today. 2019;27:73–98.

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Ngobili TA, Daniele MA. Nanoparticles and direct immunosuppression. Exp Biol Med. 2016;241(10):1064–73.

    CAS  Google Scholar 

  69. Khelghati N, et al. The importance of co-delivery of nanoparticle-siRNA and anticancer agents in cancer therapy. Chem Biol Drug Des. 2021;97(4):997–1015.

    CAS  PubMed  Google Scholar 

  70. Pecot CV, et al. RNA interference in the clinic: challenges and future directions. Nat Rev Cancer. 2011;11(1):59–67.

    CAS  PubMed  Google Scholar 

  71. Chen Y, et al. Blood clot scaffold loaded with liposome vaccine and siRNAs Targeting PD-L1 and TIM-3 for effective DC activation and cancer immunotherapy. ACS Nano. 2023;17(1):760–74.

    CAS  PubMed  Google Scholar 

  72. Strop P, et al. Location matters: site of conjugation modulates stability and pharmacokinetics of antibody drug conjugates. Chem Biol. 2013;20(2):161–7.

    CAS  PubMed  Google Scholar 

  73. Mitchell MJ, et al. Engineering precision nanoparticles for drug delivery. Nat Rev Drug Discov. 2021;20(2):101–24.

    CAS  PubMed  Google Scholar 

  74. Lee H, et al. (64)Cu-MM-302 positron emission tomography quantifies variability of enhanced permeability and retention of nanoparticles in relation to treatment response in patients with metastatic breast cancer. Clin Cancer Res. 2017;23(15):4190–202.

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Li HJ, et al. Stimuli-responsive clustered nanoparticles for improved tumor penetration and therapeutic efficacy. Proc Natl Acad Sci USA. 2016;113(15):4164–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Rodallec A, et al. Turning cold tumors into hot tumors: harnessing the potential of tumor immunity using nanoparticles. Expert Opin Drug Metab Toxicol. 2018;14(11):1139–47.

    CAS  PubMed  Google Scholar 

  77. Chen G, et al. Nanochemistry and nanomedicine for nanoparticle-based diagnostics and therapy. Chem Rev. 2016;116(5):2826–85.

    CAS  PubMed  Google Scholar 

  78. Davis ME, Chen ZG, Shin DM. Nanoparticle therapeutics: an emerging treatment modality for cancer. Nat Rev Drug Discov. 2008;7(9):771–82.

    CAS  PubMed  Google Scholar 

  79. Toy R, Roy K. Engineering nanoparticles to overcome barriers to immunotherapy. Bioeng Transl Med. 2016;1(1):47–62.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Huang Y, Zeng J. Recent development and applications of nanomaterials for cancer immunotherapy. Nanotechnol Rev. 2020;9(1):367–84.

    Google Scholar 

  81. Shen S, et al. Immunosuppressive nanoparticles for management of immune-related adverse events in liver. ACS Nano. 2021;15(5):9111–25.

    CAS  PubMed  Google Scholar 

  82. Xiong Y, Wang Y, Tiruthani K. Tumor immune microenvironment and nano-immunotherapeutics in colorectal cancer. Nanomed Nanotechnol Biol Med. 2019;21:102034.

    CAS  Google Scholar 

  83. Jurj A, et al. The new era of nanotechnology, an alternative to change cancer treatment. Drug Des Devel Ther. 2017;11:2871–90.

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Gu Z, et al. Liposome-based drug delivery systems in cancer immunotherapy. Pharmaceutics. 2020;12(11):1054.

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Lee ES, et al. Recent advances in polymeric nanomedicines for cancer immunotherapy. Adv Healthcare Mater. 2019;8(4):1801320.

    Google Scholar 

  86. Zeng Q, et al. Tailoring polymeric hybrid micelles with lymph node targeting ability to improve the potency of cancer vaccines. Biomaterials. 2017;122:105–13.

    CAS  PubMed  Google Scholar 

  87. Kiaie SH, et al. Axial pharmaceutical properties of liposome in cancer therapy: recent advances and perspectives. Int J Pharm. 2020;581: 119269.

    CAS  PubMed  Google Scholar 

  88. Ragelle H, et al. Chitosan nanoparticles for SiRNA delivery in vitro, in SiRNA Delivery Methods. Berlinp: Springer; 2016.

    Google Scholar 

  89. Zakeri A, et al. Polyethylenimine-based nanocarriers in co-delivery of drug and gene: a developing horizon. Nano Rev Exp. 2018;9(1):1488497.

    PubMed  PubMed Central  Google Scholar 

  90. Esmaily M, et al. Blockade of CTLA-4 increases anti-tumor response inducing potential of dendritic cell vaccine. J Control Rel. 2020;326:63–74.

    CAS  Google Scholar 

  91. Jiao Q, et al. Immunomodulation of nanoparticles in nanomedicine applications. BioMed Res Int. 2014. https://doi.org/10.1155/2014/426028.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Ou W, et al. Regulatory T cell-targeted hybrid nanoparticles combined with immuno-checkpoint blockage for cancer immunotherapy. J Control Release. 2018;281:84–96.

    CAS  PubMed  Google Scholar 

  93. He C, Lu J, Lin W. Hybrid nanoparticles for combination therapy of cancer. J Control Release. 2015;219:224–36.

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Wan WJ, et al. Doxorubicin and siRNA-PD-L1 co-delivery with T7 modified ROS-sensitive nanoparticles for tumor chemoimmunotherapy. Int J Pharm. 2019;566:731–44.

    CAS  PubMed  Google Scholar 

  95. Wang Y, et al. IR792-MCN@ZIF-8-PD-L1 siRNA drug delivery system enhances photothermal immunotherapy for triple-negative breast cancer under near-infrared laser irradiation. J Nanobiotechnol. 2022;20(1):96.

    Google Scholar 

  96. Hu Q, et al. Combinational chemoimmunotherapy for breast cancer by codelivery of doxorubicin and PD-L1 siRNA Using a PAMAM-incorporated liposomal nanoplatform. ACS Appl Mater Interfaces. 2022;14(7):8782–92.

    CAS  PubMed  Google Scholar 

  97. Camorani S, et al. Aptamer-functionalized nanoparticles mediate PD-L1 siRNA delivery for effective gene silencing in triple-negative breast cancer cells. Pharmaceutics. 2022. https://doi.org/10.3390/pharmaceutics14102225.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Gong T, et al. A nanodrug incorporating siRNA PD-L1 and Birinapant for enhancing tumor immunotherapy. Biomaterials Sci. 2021;9(23):8007–18.

    CAS  Google Scholar 

  99. Pacheco-Torres J, et al. PD-L1 siRNA theranostics with a dextran nanoparticle highlights the importance of nanoparticle delivery for effective tumor PD-L1 Downregulation. Front Oncol. 2021. https://doi.org/10.3389/fonc.2020.614365.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Chen M, Chen M, He J. Cancer cell membrane cloaking nanoparticles for targeted co-delivery of doxorubicin and PD-L1 siRNA. Artificial Cells Nanomed Biotechnol. 2019;47(1):1635–41.

    CAS  Google Scholar 

  101. Li C, Han X. Melanoma cancer immunotherapy using PD-L1 siRNA and imatinib promotes cancer-immunity cycle. Pharm Res. 2020;37(6):109.

    PubMed  Google Scholar 

  102. Wang D, et al. Acid-activatable versatile micelleplexes for PD-L1 blockade-enhanced cancer photodynamic immunotherapy. Nano Lett. 2016;16(9):5503–13.

    CAS  PubMed  Google Scholar 

  103. Dai L, et al. Size/charge changeable acidity-responsive micelleplex for photodynamic-improved PD-L1 immunotherapy with enhanced tumor penetration. Adv Func Mater. 2018;28(18):1707249.

    Google Scholar 

  104. Bastaki S, et al. Codelivery of STAT3 and PD-L1 siRNA by hyaluronate-TAT trimethyl/thiolated chitosan nanoparticles suppresses cancer progression in tumor-bearing mice. Life Sci. 2021;266: 118847.

    CAS  PubMed  Google Scholar 

  105. Li X, et al. Liposomal Co-delivery of PD-L1 siRNA/Anemoside B4 for enhanced combinational immunotherapeutic effect. ACS Appl Mater Interfaces. 2022;14(25):28439–54.

    CAS  PubMed  Google Scholar 

  106. Gao L, et al. Construction of PD-L1-siRNA and IL-2 DNA loading PEI lipid nanoparticles in activating T cells and treating lung cancer. AIP Adv. 2021;11(4): 045201.

    CAS  Google Scholar 

  107. Luo X, et al. Folic acid-functionalized polyethylenimine superparamagnetic iron oxide nanoparticles as theranostic agents for magnetic resonance imaging and PD-L1 siRNA delivery for gastric cancer. Int J Nanomed. 2017;12:5331.

    CAS  Google Scholar 

  108. Huang K-W, et al. Highly efficient and tumor-selective nanoparticles for dual-targeted immunogene therapy against cancer. Sci Adv. 2020. https://doi.org/10.1126/sciadv.aax5032.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Yoo B, et al. RNAi-Mediated PD-L1 inhibition for pancreatic cancer immunotherapy. Sci Rep. 2019;9(1):4712.

    PubMed  PubMed Central  Google Scholar 

  110. Ruan H, et al. A dual-bioresponsive drug-delivery depot for combination of epigenetic modulation and immune checkpoint blockade. Adv Mater. 2019;31(17):1806957.

    Google Scholar 

  111. Wang C, et al. Enhanced cancer immunotherapy by microneedle patch-assisted delivery of anti-PD1 antibody. Nano Lett. 2016;16(4):2334–40.

    CAS  PubMed  Google Scholar 

  112. Wang C, et al. Inflammation-triggered cancer immunotherapy by programmed delivery of CpG and anti-PD1 antibody. Adv Mater. 2016;28(40):8912–20.

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Su Z, et al. Codelivery of Anti-PD-1 antibody and paclitaxel with matrix metalloproteinase and pH dual-sensitive micelles for enhanced tumor chemoimmunotherapy. Small. 2020;16(7): e1906832.

    PubMed  Google Scholar 

  114. Mi Y, et al. A dual immunotherapy nanoparticle improves T-cell activation and cancer immunotherapy. Adv Mater. 2018;30(25):1706098.

    Google Scholar 

  115. Li Y, et al. A dendritic cell-like biomimetic nanoparticle enhances T cell activation for breast cancer immunotherapy. Chem Sci. 2022;13(1):105–10.

    Google Scholar 

  116. Kosmides AK, et al. Dual targeting nanoparticle stimulates the immune system to inhibit tumor growth. ACS Nano. 2017;11(6):5417–29.

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Reda M, et al. Development of a nanoparticle-based immunotherapy targeting PD-L1 and PLK1 for lung cancer treatment. Nat Commun. 2022;13(1):4261.

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Zhang P, et al. Therapeutic targeting of tumor-associated myeloid cells synergizes with radiation therapy for glioblastoma. Proc Natl Acad Sci. 2019;116(47):23714–23.

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Goodwin TJ, et al. Liver specific gene immunotherapies resolve immune suppressive ectopic lymphoid structures of liver metastases and prolong survival. Biomaterials. 2017;141:260–71.

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Miao L, et al. Transient and local expression of chemokine and immune checkpoint traps to treat pancreatic cancer. ACS Nano. 2017;11(9):8690–706.

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Gautam A, et al. Plant viral nanoparticle conjugated with anti-PD-1 peptide for ovarian cancer immunotherapy. Int J Mol Sci. 2021. https://doi.org/10.3390/ijms22189733.

    Article  PubMed  PubMed Central  Google Scholar 

  122. Luo L, et al. Sustained release of anti-PD-1 peptide for perdurable immunotherapy together with photothermal ablation against primary and distant tumors. J Control Release. 2018;278:87–99.

    CAS  PubMed  Google Scholar 

  123. Wu L, et al. Intravenous Delivery of RNA Encoding Anti-PD-1 Human Monoclonal Antibody for Treating Intestinal Cancer. J Cancer. 2022;13(2):579–88.

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Li SY, et al. Restoring anti-tumor functions of T cells via nanoparticle-mediated immune checkpoint modulation. J Control Rel. 2016;231:17–28.

    CAS  Google Scholar 

  125. Lei C, et al. Local release of highly loaded antibodies from functionalized nanoporous support for cancer immunotherapy. J Am Chem Soc. 2010;132(20):6906–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Rahimian S, et al. Polymeric microparticles for sustained and local delivery of antiCD40 and antiCTLA-4 in immunotherapy of cancer. Biomaterials. 2015;61:33–40.

    CAS  PubMed  Google Scholar 

  127. Sun J-J, et al. Programmable co-delivery of the immune checkpoint inhibitor NLG919 and chemotherapeutic doxorubicin via a redox-responsive immunostimulatory polymeric prodrug carrier. Acta Pharmacol Sin. 2017;38(6):823–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Song C, et al. Improved anti-hepatocellular carcinoma effect by enhanced Co-delivery of Tim-3 siRNA and sorafenib via multiple pH triggered drug-eluting nanoparticles. Materials Today Bio. 2022;16: 100350.

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Chen C, et al. Asynchronous blockade of PD-L1 and CD155 by polymeric nanoparticles inhibits triple-negative breast cancer progression and metastasis. Biomaterials. 2021;275: 120988.

    CAS  PubMed  Google Scholar 

  130. Zhang X, et al. PD-1 blockade cellular vesicles for cancer immunotherapy. Adv Mater. 2018;30(22):1707112.

    Google Scholar 

  131. Ye Y, et al. Synergistic transcutaneous immunotherapy enhances antitumor immune responses through delivery of checkpoint inhibitors. ACS Nano. 2016;10(9):8956–63.

    CAS  PubMed  Google Scholar 

  132. Galstyan A, et al. Blood–brain barrier permeable nano immunoconjugates induce local immune responses for glioma therapy. Nat Commun. 2019;10(1):1–13.

    Google Scholar 

  133. Kwak SY, et al. PLGA Nanoparticles codelivering siRNAs against programmed cell death protein-1 and its ligand gene for suppression of colon tumor growth. Mol Pharm. 2019;16(12):4940–53.

    CAS  PubMed  Google Scholar 

  134. Shariati M, et al. Aerosolization of nanotherapeutics as a newly emerging treatment regimen for peritoneal carcinomatosis. Cancers. 2019. https://doi.org/10.3390/cancers11070906.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Huang X, et al. Carrier-free multifunctional nanomedicine for intraperitoneal disseminated ovarian cancer therapy. J Nanobiotechnology. 2022;20(1):93.

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Ornella MSC, et al. Immunotherapy for peritoneal carcinomatosis: challenges and prospective outcomes. Cancers. 2023. https://doi.org/10.3390/cancers15082383.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Rudd CE, Taylor A, Schneider H. CD28 and CTLA-4 coreceptor expression and signal transduction. Immunol Rev. 2009;229(1):12–26.

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252–64.

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Alsaab HO, et al. PD-1 and PD-L1 checkpoint signaling inhibition for cancer immunotherapy: mechanism, combinations, and clinical outcome. Front Pharmacol. 2017;8:561.

    PubMed  PubMed Central  Google Scholar 

  140. Chen L, Flies DB. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol. 2013;13(4):227–42.

    PubMed  PubMed Central  Google Scholar 

  141. Ramsay AG. Immune checkpoint blockade immunotherapy to activate anti-tumour T-cell immunity. Br J Haematol. 2013;162(3):313–25.

    CAS  PubMed  Google Scholar 

  142. Butt A, Mills K. Immunosuppressive networks and checkpoints controlling antitumor immunity and their blockade in the development of cancer immunotherapeutics and vaccines. Oncogene. 2014;33(38):4623–31.

    CAS  PubMed  Google Scholar 

  143. Webster RM. The immune checkpoint inhibitors: where are we now? Nature reviews. Drug discovery. 2014;13(12):883.

    CAS  PubMed  Google Scholar 

  144. Ghassemi AH, et al. Controlled release of octreotide and assessment of peptide acylation from poly (D, L-lactide-co-hydroxymethyl glycolide) compared to PLGA microspheres. Pharm Res. 2012;29(1):110–20.

    CAS  PubMed  Google Scholar 

  145. Lim M, et al. Current state of immunotherapy for glioblastoma. Nat Rev Clin Oncol. 2018;15(7):422–42.

    CAS  PubMed  Google Scholar 

  146. Wainwright DA, et al. Durable therapeutic efficacy utilizing combinatorial blockade against IDO, CTLA-4, and PD-L1 in mice with brain tumors. Clin Cancer Res. 2014;20(20):5290–301.

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Belcaid Z, et al. Focal radiation therapy combined with 4–1BB activation and CTLA-4 blockade yields long-term survival and a protective antigen-specific memory response in a murine glioma model. PLoS ONE. 2014;9(7): e101764.

    PubMed  PubMed Central  Google Scholar 

  148. Villaseñor R, et al. Trafficking of endogenous immunoglobulins by endothelial cells at the blood-brain barrier. Sci Rep. 2016;6:25658.

    PubMed  PubMed Central  Google Scholar 

  149. Reardon DA, et al. Glioblastoma eradication following immune checkpoint blockade in an orthotopic. Immunocompetent Model Cancer Immunol Res. 2016;4(2):124–35.

    CAS  PubMed  Google Scholar 

  150. Brennan CW, et al. The somatic genomic landscape of glioblastoma. Cell. 2014;157(3):753.

    CAS  Google Scholar 

  151. Fransen MF, et al. Controlled local delivery of CTLA-4 blocking antibody induces CD8+ T-cell–dependent tumor eradication and decreases risk of toxic side effects. Clin Cancer Res. 2013;19(19):5381–9.

    CAS  PubMed  Google Scholar 

  152. Lei C, et al. Characterization of functionalized nanoporous supports for protein confinement. Nanotechnology. 2006;17(22):5531.

    CAS  PubMed  Google Scholar 

  153. Lei C, et al. Entrapping enzyme in a functionalized nanoporous support. J Am Chem Soc. 2002;124(38):11242–3.

    CAS  PubMed  Google Scholar 

  154. Xu J, et al. Near-infrared-triggered photodynamic therapy with multitasking upconversion nanoparticles in combination with checkpoint blockade for immunotherapy of colorectal cancer. ACS Nano. 2017;11(5):4463–74.

    CAS  PubMed  Google Scholar 

  155. Curran MA, et al. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci. 2010;107(9):4275–80.

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Tian M, et al. The PD-1/PD-L1 inhibitory pathway is altered in pre-eclampsia and regulates T cell responses in pre-eclamptic rats. Sci Rep. 2016;6(1):1–14.

    Google Scholar 

  157. Ohaegbulam KC, et al. Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway. Trends Mol Med. 2015;21(1):24–33.

    CAS  PubMed  Google Scholar 

  158. Muenst S, et al. Expression of programmed death ligand 1 (PD-L1) is associated with poor prognosis in human breast cancer. Breast Cancer Res Treat. 2014;146(1):15–24.

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Schalper KA. PD-L1 expression and tumor-infiltrating lymphocytes: revisiting the antitumor immune response potential in breast cancer. Oncoimmunology. 2014;3(6): e29288.

    PubMed  PubMed Central  Google Scholar 

  160. Tang J, et al. Preparation of optimized lipid-coated calcium phosphate nanoparticles for enhanced in vitro gene delivery to breast cancer cells. J Mater Chem B. 2015;3(33):6805–12.

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Ma D. Enhancing endosomal escape for nanoparticle mediated siRNA delivery. Nanoscale. 2014;6(12):6415–25.

    CAS  PubMed  Google Scholar 

  162. !!! INVALID CITATION !!! {Ma, 2014 #344;Khalilzadeh, 2022 #70}.

  163. Wu Y, et al. Enhancing PD-1 gene silence in T lymphocytes by comparing the delivery performance of two inorganic nanoparticle platforms. Nanomaterials. 2019;9(2):159.

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Wang C, et al. In situ activation of platelets with checkpoint inhibitors for post-surgical cancer immunotherapy. Nature Biomedical Engineering. 2017;1(2):1–10.

    Google Scholar 

  165. Marabelle A, et al. Depleting tumor-specific Tregs at a single site eradicates disseminated tumors. J Clin Investig. 2013;123(6):2447–63.

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Krieg AM. Toll-like receptor 9 (TLR9) agonists in the treatment of cancer. Oncogene. 2008;27(2):161–7.

    CAS  PubMed  Google Scholar 

  167. Guo F, et al. CXCL12/CXCR4: a symbiotic bridge linking cancer cells and their stromal neighbors in oncogenic communication networks. Oncogene. 2016;35(7):816–26.

    CAS  PubMed  Google Scholar 

  168. Kiaie SH, et al. Immune checkpoints in targeted-immunotherapy of pancreatic cancer: new hope for clinical development. Acta Pharm Sin B. 2021;11(5):1083–97.

    CAS  PubMed  Google Scholar 

  169. Chiang C-S, et al. Combination of fucoidan-based magnetic nanoparticles and immunomodulators enhances tumour-localized immunotherapy. Nat Nanotechnol. 2018;13(8):746–54.

    CAS  PubMed  Google Scholar 

  170. Chen X-J, et al. Anti-PD-L1-modified and ATRA-loaded nanoparticles for immuno-treatment of oral dysplasia and oral squamous cell carcinoma. Nanomedicine. 2020;15(10):951–68.

    CAS  PubMed  Google Scholar 

  171. Yu J, et al. Myeloid-derived suppressor cells suppress antitumor immune responses through IDO expression and correlate with lymph node metastasis in patients with breast cancer. J Immunol. 2013;190(7):3783–97.

    CAS  PubMed  Google Scholar 

  172. Cheong JE, Sun L. Targeting the IDO1/TDO2–KYN–AhR pathway for cancer immunotherapy–challenges and opportunities. Trends Pharmacol Sci. 2018;39(3):307–25.

    CAS  PubMed  Google Scholar 

  173. Xu X, Lu H, Lee R. Near infrared light triggered photo/immuno-therapy toward cancers. Front Bioeng Biotechnol. 2020;8:488.

    PubMed  PubMed Central  Google Scholar 

  174. Meng X, et al. Photothermal/photodynamic therapy with immune-adjuvant liposomal complexes for effective gastric cancer therapy. Part Part Systems Charact. 2019. https://doi.org/10.1002/ppsc.201900015.

    Article  Google Scholar 

  175. Craik DJ, et al. The future of peptide-based drugs. Chem Biol Drug Des. 2013;81(1):136–47.

    CAS  PubMed  Google Scholar 

  176. Liang C, et al. Emerging nanomedicine approaches fighting tumor metastasis: animal models, metastasis-targeted drug delivery, phototherapy, and immunotherapy. Chem Soc Rev. 2016;45(22):6250–69.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not Applicable.

Funding

Not Applicable.

Author information

Authors and Affiliations

Authors

Contributions

SHK designed the work. SHK, HSS, JS and MA collected data and wrote the manuscript. MSN, and MS, checked and revised the manuscript. SHK and MS revised the article critically. All of the authors have read and approved the final manuscript.

Corresponding authors

Correspondence to Seyed Hossein Kiaie or Mohammad Sheibani.

Ethics declarations

Ethics approval and consent to participate

Not Applicable.

Consent for publication

Not Applicable.

Competing interests

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

The original online version of this article has been revised: the graphical abstract is updated

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kiaie, S.H., Salehi-Shadkami, H., Sanaei, M.J. et al. Nano-immunotherapy: overcoming delivery challenge of immune checkpoint therapy. J Nanobiotechnol 21, 339 (2023). https://doi.org/10.1186/s12951-023-02083-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12951-023-02083-y

Keywords